Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941676

RESUMO

Chronic inflammatory diseases like rheumatoid arthritis are characterized by a deficit in fully functional regulatory T cells. DNA-methylation inhibitors have previously been shown to promote regulatory T cell responses and, in the present study, we evaluated their potential to ameliorate chronic and acute animal models of rheumatoid arthritis. Of the drugs tested, decitabine was the most effective, producing a sustained therapeutic effect that was dependent on indoleamine 2,3-dioxygenase (IDO) and was associated with expansion of induced regulatory T cells, particularly at the site of disease activity. Treatment with decitabine also caused apoptosis of Th1 and Th17 cells in active arthritis in a highly selective manner. The molecular basis for this selectivity was shown to be ENT1, a nucleoside transporter, which facilitates intracellular entry of the drug and is up-regulated on effector T cells during active arthritis. It was further shown that short-term treatment with decitabine resulted in the generation of a population of regulatory T cells that were able to suppress arthritis upon adoptive transfer. In summary, a therapeutic approach using an approved drug is described that treats active inflammatory disease effectively and generates robust regulatory T cells with the IDO-dependent capacity to maintain remission.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Decitabina/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/genética , Transportador Equilibrativo 1 de Nucleosídeo/imunologia , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Indução de Remissão , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th17/citologia , Células Th17/imunologia
2.
Cancer Treat Res Commun ; 26: 100293, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33385735

RESUMO

BACKGROUND: Glioblastoma is the most common primary malignant brain tumor in adults. Previous studies have suggested that CRP (C-reactive protein) could serve as a biomarker candidate as well as a prognostic factor in glioblastoma patients, and we here further investigate its potential role. MATERIALS AND METHODS: Publicly available datasets were used to compare gene expression between brain samples from glioblastoma patients and non-tumor tissue. The structure of CRP was compared between humans and rats. Glioblastoma cells from humans and rats were stained with anti-CRP. Fischer 344 rats were inoculated with syngeneic glioblastoma cells pre-coated with anti-CRP, and survival was monitored. CRP concentration in rats carrying glioblastoma was followed. RESULTS: CRP was upregulated on one locus on gene level in glioblastoma tissue as compared to non-tumor brain tissue, but not in glioma stem cells as compared to neural stem cells. The structure of the CRP protein was a characteristic pentamer in both humans and rats. Both human and rat glioblastoma cells were clearly positive for anti-CRP staining. Pre-coating of glioblastoma cells with anti-CRP antibodies did not affect survival in rats with intracranial tumors. Serum levels of CRP increased during tumor progression but did not reach significantly different levels. CONCLUSIONS: Both human and rat glioblastoma cells could be stained with anti-CRP antibodies in vitro. In a syngeneic glioblastoma rat model we could see an increase in serum CRP during tumor progression, but coating glioblastoma cells with anti-CRP antibodies did not provide any survival change for the animals.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Encefálicas/diagnóstico , Encéfalo/patologia , Proteína C-Reativa/análise , Glioblastoma/diagnóstico , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Proteína C-Reativa/metabolismo , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Progressão da Doença , Feminino , Glioblastoma/sangue , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Prognóstico , Ratos , Análise de Sobrevida
3.
Histol Histopathol ; 35(8): 871-886, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32022242

RESUMO

Glioblastoma multiforme (GBM) is an aggressive primary brain malignancy with a very poor prognosis. Researchers employ animal models to develop potential therapies. It is important that these models have clinical relevance. This means that old models, propagated for decades in cultures, should be questioned. Parameters to be evaluated include whether animals are immune competent or not, the infiltrative growth pattern of the tumor, tumor volume resulting in symptoms and growth rate. We here describe the growth pattern of an experimental glioblastoma model in detail with GFP positive glioblastoma cells in fully immune competent animals and study tumor growth rate and tumor mass as a function of time from inoculation. We were able to correlate findings made with classical immunohistochemistry and MR findings. The tumor growth rate was fitted by a Gompertz function. The model predicted the time until onset of symptoms for 5000 inoculated cells to 18.7±0.4 days, and the tumor mass at days 10 and 14, which are commonly used as the start of treatment in therapeutic studies, were 5.97±0.62 mg and 29.1±3.0 mg, respectively. We want to raise the question regarding the clinical relevance of the outline of glioblastoma experiments, where treatment is often initiated at a very early stage. The approach presented here could potentially be modified to gain information also from other tumor models.


Assuntos
Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioblastoma/patologia , Transplante de Neoplasias/métodos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ratos , Ratos Endogâmicos F344
4.
Oncotarget ; 10(55): 5703-5712, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31620245

RESUMO

PURPOSE: The complement system has recently sparked more interest in cancer research. The classical pathway is initiated by activation of the C1 complex, which irreversibly can be bound to and inhibited by C1-INH. We have previously shown that C1-INH is upregulated in human glioblastoma (astrocytoma grade IV) on both gene and protein level. We here examine whether the complement system seems to play a role also in pancreatic cancer. TECHNIQUE AND RESULTS: We performed an expression analysis of complement associated genes in 36 pancreatic ductal adenocarcinoma tumors and matching normal pancreatic tissue samples from pancreatic cancer patients (data from the publicly available database GSE15471). C1-INH was significantly upregulated in the pancreatic cancer tissue. None of the downstream components of the cascade were significantly upregulated in the cancer samples as compared to the control samples, which is the same pattern as we found in glioblastoma. GO analysis showed that membrane attack complex came up as the second most significantly associated cellular component. Analyzing gene expression of C1-INH in the pancreatic cancer cell lines from primary tumors versus metastatic tumor revealed no difference for the two mRNA transcripts (GSE59357). INTERPRETATION: Analysis of gene expression of complement related genes shows an upregulation of C1-INH and a downregulation of downstream components. This could suggest that C1-INH plays a role also in pancreatic cancer.

5.
Oncotarget ; 9(9): 8391-8399, 2018 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-29492202

RESUMO

Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor, and available experimental and routine therapies result in limited survival benefits. A vulnerability of GBM cells to catastrophic vacuolization and cell death, a process termed methuosis, induced by Vacquinol-1 (VQ-1) has been described earlier. In the present study, we investigate the efficacy of VQ-1 treatment in two syngeneic rat GBM models, RG2 and NS1. VQ-1 treatment affected growth of both RG2 and NS1 cells in vitro. Intracranially, significant reduction in RG2 tumor size was observed, although no effect was seen on overall survival. No survival advantage or effect on tumor size was seen in animals carrying the NS1 models compared to untreated controls. Furthermore, immunological staining of FOXP3, CD4 and CD8 showed no marked difference in immune cell infiltrate in tumor environment following treatment. Taken together, a survival advantage of VQ-1 treatment alone could not be demonstrated here, even though some effect upon tumor size was seen. Staining for immune cell markers did not indicate that VQ-1 either reduced or increased host anti-tumor immune response.

6.
Oncotarget ; 9(100): 37421-37428, 2018 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-30647842

RESUMO

PURPOSE: Glioblastoma multiforme (GBM) or astrocytoma grade IV is the most common type of primary brain tumor in adults. In the present study, we investigate the role of the complement system in the glioblastoma situation in an experimental model, since we have previously been able to show a blockade of this system in the glioblastoma setting. TECHNIQUE AND RESULTS: A GFP-positive glioblastoma cell line was used to induce glioblastomas subcutaneously in rats (n=42). Antibodies against C1-Inactivator (C1-IA) were used to try to re-activate the complement system. We were able to demonstrate an increased survival in rats treated with anti-C1-IA with an intratumoral route, and we could establish the same the results in a second series. Serum analyses revealed decreased levels of IL-1b and GM-CSF in animals 24 days after tumor cell inoculation in the anti-C1-IA group when compared to controls. Immunohistochemistry revealed decreased expression of C1-IA following treatment. INTERPRETATION: These results are in line with our previous work showing an upregulation of C1-IA, which is able to block the classical complement pathway, in glioblastomas. Treatment with antibodies against C1-IA seems to be beneficial in the glioblastoma situation, and no side effects could be seen in our experiments.

7.
PLoS One ; 12(9): e0183086, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28880870

RESUMO

BACKGROUND: Glioblastoma is the most common and aggressive type of primary brain tumor in adults. A key problem is the capacity of glioma cells to inactivate the body's immune response. The complement system acts as a functional bridge between the innate and adaptive immune response. Still, the role of the complement system has almost been forgotten in glioma research. In our present study, we hypothesize that C1 inactivator (C1-IA) is upregulated in astrocytoma grade IV, and that its inhibition of the complement system has beneficial effects upon survival. METHODS AND RESULTS: We have explored this hypothesis both on gene and protein levels and found an upregulation of C1-IA in human glioblastoma cells using data from a publicly available database and our own mRNA material from glioblastoma patients. Furthermore, we demonstrated the presence of C1-IA by using immunohistochemistry on glioma cells from both humans and rats in vitro. Finally, we could demonstrate a significantly increased survival in vivo in animals inoculated intracerebrally with glioma cells pre-coated with C1-IA antibodies as compared to control animals. CONCLUSIONS: Our findings indicate that overexpression of C1-IA is present in glioblastomas. This could be demonstrated both at the gene level from patients with glioblastoma, on mRNA level and with immunohistochemistry. Treatment with antibodies against C1-IA had beneficial effects on survival when tested in vivo.


Assuntos
Astrocitoma/metabolismo , Proteína Inibidora do Complemento C1/metabolismo , Glioblastoma/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Feminino , Glioma/metabolismo , Humanos , Imuno-Histoquímica , Gravidez , Ratos
8.
Anticancer Res ; 36(11): 5751-5755, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27793896

RESUMO

BACKGROUND: Inositol trispyrophosphate (ITPP) has been shown to reduce tumour growth in different animal cancer models, as well as of human U87 glioma cells grafted onto chick chorioallantoic membrane (CAM). The aim of this study was to establish whether ITPP crosses the blood-brain barrier and whether it halts the growth of RG2 glioblastoma tumour. MATERIALS AND METHODS: A model comprising of Fischer 344 rats was chosen and RG2 cells were implanted either intracranially, or subcutaneously on the left hind leg, and the animals were treated with ITPP either intraperitoneally, intravenously or both routes combined. Overall survival was then calculated. RESULTS: No prolonged survival was seen in animals treated with ITPP. The route of ITPP administration did not affect outcome. CONCLUSION: ITPP had no favourable effect upon survival in our animal model with RG2 glioblastoma tumours in Fischer 344 rats.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Fosfatos de Inositol/uso terapêutico , Animais , Feminino , Ratos , Ratos Endogâmicos F344
9.
Proc Natl Acad Sci U S A ; 111(9): 3538-43, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24550449

RESUMO

We hypothesized that key signaling pathways of glioma genesis might enable the molecular classification of gliomas. Gene coexpression modules around epidermal growth factor receptor (EGFR) (EM, 29 genes) or platelet derived growth factor receptor A (PDGFRA) (PM, 40 genes) in gliomas were identified. Based on EM and PM expression signatures, nonnegative matrix factorization reproducibly clustered 1,369 adult diffuse gliomas WHO grades II-IV from four independent databases generated in three continents, into the subtypes (EM, PM and EM(low)PM(low) gliomas) in a morphology-independent manner. Besides their distinct patterns of genomic alterations, EM gliomas were associated with higher age at diagnosis, poorer prognosis, and stronger expression of neural stem cell and astrogenesis genes. Both PM and EM(low)PM(low) gliomas were associated with younger age at diagnosis and better prognosis. PM gliomas were enriched in the expression of oligodendrogenesis genes, whereas EM(low)PM(low) gliomas were enriched in the signatures of mature neurons and oligodendrocytes. The EM/PM-based molecular classification scheme is applicable to adult low-grade and high-grade diffuse gliomas, and outperforms existing classification schemes in assigning diffuse gliomas to subtypes with distinct transcriptomic and genomic profiles. The majority of the EM/PM classifiers, including regulators of glial fate decisions, have not been extensively studied in glioma biology. Subsets of these classifiers were coexpressed in mouse glial precursor cells, and frequently amplified or lost in an EM/PM glioma subtype-specific manner, resulting in somatic copy number alteration-dependent gene expression that contributes to EM/PM signatures in glioma samples. EM/PM-based molecular classification provides a molecular diagnostic framework to expedite the search for new glioma therapeutic targets.


Assuntos
Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glioma/classificação , Glioma/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/genética , Fatores Etários , Animais , China , Análise por Conglomerados , Receptores ErbB/genética , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Camundongos , Células-Tronco Neurais/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Prognóstico , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
10.
PLoS One ; 9(1): e87281, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489888

RESUMO

Increased PDGFRA signaling is an essential pathogenic factor in many subtypes of gliomas. In this context the cell surface expression of PDGFRA is an important determinant of ligand sensing in the glioma microenvironment. However, the regulation of spatial distribution of PDGFRA in glioma cells remains poorly characterized. Here, we report that cell surface PDGFRA expression in gliomas is negatively regulated by an ERK-dependent mechanism, resulting in reduced proliferation of glioma cells. Glioma tumor tissues and their corresponding cell lines were isolated from 14 patients and analyzed by single-cell imaging and flow cytometry. In both cell lines and their corresponding tumor samples, glioma cell proliferation correlated with the extent of surface expression of PDGFRA. High levels of surface PDGFRA also correlated to high tubulin expression in glioma tumor tissue in vivo. In glioma cell lines, surface PDGFRA declined following treatment with inhibitors of tubulin, actin and dynamin. Screening of a panel of small molecule compounds identified the MEK inhibitor U0126 as a potent inhibitor of surface PDGFRA expression. Importantly, U0126 inhibited surface expression in a reversible, dose- and time-dependent manner, without affecting general PDGFRA expression. Treatment with U0126 resulted in reduced co-localization between PDGFRA and intracellular trafficking molecules e.g. clathrin, RAB11 and early endosomal antigen-1, in parallel with enhanced co-localization between PDGFRA and the Golgi cisternae maker, Giantin, suggesting a deviation of PDGFRA from the endosomal trafficking and recycling compartment, to the Golgi network. Furthermore, U0126 treatment in glioma cells induced an initial inhibition of ERK1/2 phosphorylation, followed by up-regulated ERK1/2 phosphorylation concomitant with diminished surface expression of PDGFRA. Finally, down-regulation of surface PDGFRA expression by U0126 is concordant with reduced glioma cell proliferation. These findings suggest that manipulation of spatial expression of PDGFRA can potentially be used to combat gliomas.


Assuntos
Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioma/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Butadienos/farmacologia , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Células Tumorais Cultivadas , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
11.
PLoS One ; 8(8): e71981, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991016

RESUMO

BACKGROUND: Coping with the immune rejection of allotransplants or autologous cells in patients with an active sensitization towards their autoantigens and autoimmunity presently necessitates life-long immune suppressive therapy acting on the immune system as a whole, which makes the patients vulnerable to infections and increases their risk of developing cancer. New technologies to induce antigen selective long-lasting immunosuppression or immune tolerance are therefore much needed. METHODOLOGY/PRINCIPAL FINDINGS: The DNA demethylating agent Zebularine, previously demonstrated to induce expression of the genes for the immunosuppressive enzymes indolamine-2,3-deoxygenase-1 (IDO1) and kynureninase of the kynurenine pathway, is tested for capacity to suppress rejection of allotransplants. Allogeneic pancreatic islets from Lewis rats were transplanted under the kidney capsule of Fischer rats previously made diabetic by a streptozotocin injection (40 mg/kg). One group was treated with Zebularine (225 mg/kg) daily for 14 days from day 6 or 8 after transplantation, and a control group received no further treatment. Survival of the transplants was monitored by blood sugar measurements. Rats, normoglycemic for 90 days after allografting, were subjected to transplant removal by nephrectomy to confirm whether normoglycemia was indeed due to a surviving insulin producing transplant, or alternatively was a result of recovery of pancreatic insulin production in some toxin-treated rats. Of 9 Zebularine treated rats, 4 were still normoglycemic after 90 days and became hyperglycemic after nephrectomy. The mean length of normoglycemia in the Zebularine group was 67±8 days as compared to 14±3 days in 9 controls. Seven rats (2 controls and 5 Zebularine treated) were normoglycemic at 90 days due to pancreatic recovery as demonstrated by failure of nephrectomy to induce hyperglycemia. CONCLUSIONS/SIGNIFICANCE: Zebularine treatment in vivo induces a long-lasting suppression of the immune destruction of allogeneic pancreatic islets resulting in protection of allograft function for more than 10 weeks after end of treatment.


Assuntos
Citidina/análogos & derivados , Diabetes Mellitus Experimental/cirurgia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas/métodos , Animais , Glicemia/metabolismo , Citidina/farmacologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/imunologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Transplante das Ilhotas Pancreáticas/imunologia , Masculino , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/efeitos dos fármacos , Baço/metabolismo , Fatores de Tempo , Transplante Homólogo , Resultado do Tratamento
12.
PLoS One ; 8(4): e61556, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23630597

RESUMO

Signaling of platelet derived growth factor receptor alpha (PDGFRA) is critically involved in the development of gliomas. However, the clinical relevance of PDGFRA expression in glioma subtypes and the mechanisms of PDGFRA expression in gliomas have been controversial. Under the supervision of morphological diagnosis, analysis of the GSE16011 and the Repository of Molecular Brain Neoplasia Data (Rembrandt) set revealed enriched PDGFRA expression in low-grade gliomas. However, gliomas with the top 25% of PDGFRA expression levels contained nearly all morphological subtypes, which was associated with frequent IDH1 mutation, 1p LOH, 19q LOH, less EGFR amplification, younger age at disease onset and better survival compared to those gliomas with lower levels of PDGFRA expression. SNP analysis in Rembrandt data set and FISH analysis in eleven low passage glioma cell lines showed infrequent amplification of PDGFRA. Using in vitro culture of these low passage glioma cells, we tested the hypothesis of gliogenic factor dependent expression of PDGFRA in glioma cells. Fibroblast growth factor 2 (FGF2) was able to maintain PDGFRA expression in glioma cells. FGF2 also induced PDGFRA expression in glioma cells with low or non-detectable PDGFRA expression. FGF2-dependent maintenance of PDGFRA expression was concordant with the maintenance of a subset of gliogenic genes and higher rates of cell proliferation. Further, concordant expression patterns of FGF2 and PDGFRA were detected in glioma samples by immunohistochemical staining. Our findings suggest a role of FGF2 in regulating PDGFRA expression in the subset of gliomas with younger age at disease onset and longer patient survival regardless of their morphological diagnosis.


Assuntos
Neoplasias Encefálicas/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glioma/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Fator 2 de Crescimento de Fibroblastos/genética , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Glioma/mortalidade , Glioma/patologia , Humanos , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Transcriptoma
13.
Phys Med Biol ; 57(24): 8377-91, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23201928

RESUMO

75 RG2 glioma-carrying Fischer rats were treated by photon activation therapy (PAT) with monochromatic synchrotron radiation and stable thallium. Three groups were treated with thallium in combination with radiation at different energy; immediately below and above the thallium K-edge, and at 50 keV. Three control groups were given irradiation only, thallium only, or no treatment at all. For animals receiving thallium in combination with radiation to 15 Gy at 50 keV, the median survival time was 30 days, which was 67% longer than for the untreated controls (p = 0.0020) and 36% longer than for the group treated with radiation alone (not significant). Treatment with thallium and radiation at the higher energy levels were not effective at the given absorbed dose and thallium concentration. In the groups treated at 50 keV and above the K-edge, several animals exhibited extensive and sometimes contra-lateral edema, neuronal death and frank tissue necrosis. No such marked changes were seen in the other groups. The results were discussed with reference to Monte Carlo calculated electron energy spectra and dose enhancement factors.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioma/radioterapia , Fótons/uso terapêutico , Radioterapia/instrumentação , Síncrotrons , Tálio/química , Tálio/uso terapêutico , Animais , Encéfalo/citologia , Encéfalo/patologia , Encéfalo/efeitos da radiação , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Elétrons , Glioma/diagnóstico por imagem , Glioma/patologia , Masculino , Dosagem Radioterapêutica , Ratos , Ratos Endogâmicos F344 , Tomografia Computadorizada por Raios X
14.
Bioelectromagnetics ; 33(7): 535-42, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22487968

RESUMO

The question whether very weak, low frequency magnetic fields can affect biological matter is still under debate. The theoretical possibility of such an interaction is often questioned and the site of interaction in the cell is unknown. In the present study, the influence of extremely weak 60 Hz magnetic fields on the transport of Ca(2+) was studied in a biological system consisting of highly purified plasma membrane vesicles. We tested a newly proposed quantum mechanical model postulates that polarization of hydrogen nuclei can elicit a biological effect. Vesicles were exposed for half an hour at 32 °C and the calcium efflux was studied using radioactive (45) Ca(2+) as a tracer. A static magnetic field of 26 µT and time-varying magnetic fields with a frequency of 60 Hz and amplitudes between 0.6 and 6.3 µT were used. The predictions of the model, proposed by Lednev, that at a frequency of 60 Hz the biological effect under investigation would significantly be altered at the amplitudes of 1.3 and 3.9 µT could not be confirmed.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efeitos da radiação , Campos Eletromagnéticos , Modelos Biológicos , Transporte Biológico/efeitos da radiação , Canais de Cálcio/metabolismo , Espectroscopia de Ressonância Magnética , Spinacia oleracea/citologia , Spinacia oleracea/efeitos da radiação , Tempo
15.
Mol Immunol ; 51(2): 101-11, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22424783

RESUMO

IDO1 can be induced by interferon gamma (IFN-γ) in multiple cell types. We have earlier described that the DNA methyltransferase inhibitor zebularine also induces IDO1 in several rat cell clones. We now describe a synergistic induction of IDO1 expression by IFN-γ and zebularine. To elucidate the mechanism of the IDO1 induction we have studied the methylation status in the promoter region of the IDO1 gene from both human monocytic THP-1 cells and H1D2 rat colon cancer cells. Interestingly, the IDO1 promoter is hypermethylated and IFN-γ is shown to induce a significant demethylation. The synergism in effect of zebularine and IFN-γ on IDO1 expression is paralleled by a similar synergistic effect on expression of two other IFN-γ-responsive genes, the transcription factors STAT1 and IRF1 with binding sites in the IDO1 promoter region. The demonstrated synergistic activation of IDO1 expression has implications in relation to therapeutic induction of immunosuppression in autoimmunity and chronic inflammation.


Assuntos
Citidina/análogos & derivados , Epigênese Genética/genética , Expressão Gênica/efeitos dos fármacos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Interferon gama/administração & dosagem , Animais , Doenças Autoimunes/genética , Células Cultivadas , Citidina/administração & dosagem , Metilação de DNA , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética , Regiões Promotoras Genéticas , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/genética
16.
Int J Radiat Biol ; 88(3): 245-52, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22124250

RESUMO

PURPOSE: To investigate whether mobile phone radiation might affect snail nociception, employing radiofrequency (RF) electromagnetic fields (EMF) which, to our knowledge, have hitherto not been studied in a snail model. Exposure to extremely low frequency (ELF) magnetic fields has however been shown to significantly affect nociceptive responses. MATERIALS AND METHODS: In the present study, we exposed 29 land snails of the strain Helix pomatia to global system for mobile communications (GSM) EMF at 1900 MHz at the non-thermal level 48 mW/kg for 1 hour each and 29 snails were sham controls. The experiments took place during the onset of summer, with all snails being well out of hibernation. Before and after GSM or sham exposure, the snails were subjected to thermal pain by being placed on a hot plate. The reaction time for retraction from the hot plate was measured by two blinded observers. RESULTS: Comparing the reaction pattern of each snail before and after exposure, the GSM-exposed snails were less sensitive to thermal pain as compared to the sham controls, indicating that RF exposure induces a significant analgesia (Mann-Whitney p < 0.001). CONCLUSION: This study might support earlier findings, describing beneficial effects of EMF exposure upon nociception.


Assuntos
Campos Eletromagnéticos , Caracois Helix , Nociceptividade/efeitos da radiação , Terapia por Radiofrequência , Animais , Telefone Celular
17.
Neurosurgery ; 70(3): 731-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21869725

RESUMO

BACKGROUND: Viral gene therapy of malignant brain tumors has been restricted by the limited vector distribution within the tumors. Multipotent mesenchymal stromal cells (MSCs) and other precursor cells have shown tropism for gliomas, and these cells are currently being explored as potential vehicles for gene delivery in glioma gene therapy. OBJECTIVE: To investigate MSC migration in detail after intratumoral and extratumoral implantation through syngeneic and orthotopic glioma models. METHODS: Adult rat bone marrow-derived MSCs were transduced to express enhanced green fluorescent protein and implanted either directly into or at a distance from rat gliomas. RESULTS: We found no evidence of long-distance MSC migration through the intact striatum toward syngeneic D74(RG2), N32, and N29 gliomas in the ipsilateral hemisphere or across the corpus callosum to gliomas located in the contralateral hemisphere. After intratumoral injection, MSCs migrated extensively, specifically within N32 gliomas. The MSCs did not proliferate within tumors, suggesting a low risk of malignant transformation of in vivo grafted cell vectors. Using a model for surgical glioma resection, we found that intratumorally grafted MSCs migrate efficiently within glioma remnants after partial surgical resection. CONCLUSION: The findings point to limitations for the use of MSCs as vectors in glioma gene therapy, although intratumoral MSC implantation provides a dense and tumor-specific vector distribution.


Assuntos
Neoplasias Encefálicas/terapia , Movimento Celular/fisiologia , Terapia Genética/métodos , Glioma/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Proliferação de Células , Células Cultivadas , Terapia Combinada , Corpo Caloso/citologia , Corpo Estriado/citologia , Modelos Animais de Doenças , Feminino , Glioma/patologia , Glioma/cirurgia , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/genética , Masculino , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/transplante , Gravidez , Ratos , Ratos Endogâmicos F344
18.
Radiat Res ; 173(4): 433-40, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20334515

RESUMO

Single-fraction radiation therapy with 5 or 15 Gy (60)Co gamma radiation was combined with intraperitoneal injections of syngeneic interferon gamma (IFN-gamma)-transfected cells in rats with intracerebral N29 or N32 glioma tumors at days 7, 21 and 35 after inoculation. For intracerebral N29 tumors, single-fraction radiation therapy with 5 or 15 Gy had no significant effect on the survival time. Immunization with IFN-gamma-transfected N29 cells significantly increased the survival time by 61%. Single-fraction radiation therapy with 5 Gy combined with immunization increased the survival time significantly by 87% and complete remissions by 75% while with 15 Gy the survival time increased 45% with 38% complete remissions. For intracerebral N32 tumors, single-fraction radiation therapy with 15 Gy increased the survival time significantly by 20%. Immunization by itself had no significant effect with IFN-gamma-transfected N32 cells, but combined with 15 Gy single-fraction radiation therapy it increased survival time significantly by 40%, although there were no complete remissions. Based on these findings, we suggest a new therapeutic regimen for malignant glioma using single-fraction radiation therapy with a target absorbed dose of the order of 5-10 Gy combined with clinically verified immunotherapy.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Imunomodulação , Interferon gama/administração & dosagem , Animais , Neoplasias Encefálicas/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Feminino , Glioma/imunologia , Injeções Intraperitoneais , Interferon gama/genética , Interferon gama/imunologia , Masculino , Dosagem Radioterapêutica , Ratos , Ratos Endogâmicos F344 , Análise de Sobrevida , Taxa de Sobrevida , Resultado do Tratamento
19.
J Neurooncol ; 97(2): 207-15, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19809793

RESUMO

Antibodies specifically targeting tumor-associated antigens have proved to be important tools in the treatment of human cancer. A desirable target antigen should be unique to tumor cells, abundantly expressed, and readily available for antibody binding. The Ku70/80 DNA-repair protein is expressed in the nucleus of most cells; it is, however, also present on the cell surface of tumor cell lines, and antibodies binding Ku70/80 at the cell surface were recently shown to internalize into tumor cells. To evaluate the potential of Ku70/80-antigen as a therapeutic target for immunotoxins in glioblastoma multiforme, we investigated binding and localization of Ku70/80-specific antibodies in tissue samples from glioblastomas and normal human brains, and in glioma cell cultures. Furthermore, the internalization and drug-delivery capacity were evaluated by use of immunotoxicity studies. We demonstrate that Ku70/80 is localized on the cell plasma membrane of glioma cell lines, and is specifically present in human glioblastoma tissue. Antibodies bound to the Ku70/80 antigen on the cell surface of glioma cells were found to internalize via endocytosis, and shown to efficiently deliver toxins into glioblastoma cells. The data further imply that different antibodies directed against Ku70/80 possess different abilities to target the antigen, in relation to its presentation on the cell surface or intracellular localization. We conclude that Ku70/80 antigen is uniquely presented on the plasma membrane in glioblastomas, and that antibodies specific against the antigen have the capacity to selectively bind, internalize, and deliver toxins into tumor cells. These results imply that Ku70/80 is a potential target for immunotherapy of glioblastoma multiforme.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos Nucleares/metabolismo , Neoplasias Encefálicas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glioblastoma/metabolismo , Imunoterapia/métodos , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Antígenos Nucleares/imunologia , Neoplasias Encefálicas/imunologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/imunologia , Glioblastoma/imunologia , Humanos , Imuno-Histoquímica , Autoantígeno Ku , Microscopia Confocal
20.
PLoS One ; 4(12): e8484, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20041185

RESUMO

The basic concept of conditionally replicating adenoviruses (CRAD) as oncolytic agents is that progenies generated from each round of infection will disperse, infect and kill new cancer cells. However, CRAD has only inhibited, but not eradicated tumor growth in xenograft tumor therapy, and CRAD therapy has had only marginal clinical benefit to cancer patients. Here, we found that CRAD propagation and cancer cell survival co-existed for long periods of time when infection was initiated at low multiplicity of infection (MOI), and cancer cell killing was inefficient and slow compared to the assumed cell killing effect upon infection at high MOI. Excessive production of fiber molecules from initial CRAD infection of only 1 to 2% cancer cells and their release prior to the viral particle itself caused a tropism-specific receptor masking in both infected and non-infected bystander cells. Consequently, the non-infected bystander cells were inefficiently bound and infected by CRAD progenies. Further, fiber overproduction with concomitant restriction of adenovirus spread was observed in xenograft cancer therapy models. Besides the CAR-binding Ad4, Ad5, and Ad37, infection with CD46-binding Ad35 and Ad11 also caused receptor masking. Fiber overproduction and its resulting receptor masking thus play a key role in limiting CRAD functionality, but potentially promote adenovirus and host cell co-existence. These findings also give important clues for understanding mechanisms underlying the natural infection course of various adenoviruses.


Assuntos
Adenoviridae/fisiologia , Efeito Espectador , Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Infecções por Adenoviridae/metabolismo , Animais , Morte Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Camundongos , Camundongos Nus , Microscopia Confocal , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...